101
|
Zhang WF, Zhu TT, Xiong YW, Xiong AZ, Ge XY, Hu CP, Zhang Z. Negative feedback regulation between microRNA let-7g and LOX-1 mediated hypoxia-induced PASMCs proliferation. Biochem Biophys Res Commun 2017; 488:655-663. [PMID: 28108289 DOI: 10.1016/j.bbrc.2017.01.073] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 01/16/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a proliferative disorder associated with enhanced proliferation and suppressed apoptosis of pulmonary artery smooth muscle cells (PASMCs). Our lately study demonstrated that let-7g inhibited hypoxia-induced proliferation of PASMCs via repressing c-myc-Bmi-1-p16 signaling pathway. However, the upstream of let-7g has not yet been fully defined. Previous studies have shown that LOX-1, a target of let-7g, could also regulate the expression of let-7g in human aortic endothelial cells. In this present study, we aimed to investigate whether there is a negative feedback regulation between microRNA let-7g and LOX-1 in hypoxia-induced proliferation of PASMCs. METHODS SD Rats were exposed to hypoxia (10% O2, 3 weeks) to induce PH. HE staining was used to evaluate pulmonary artery remodeling. in situ hybridization and immunohistochemistry were performed to assess the expression and distribution of let-7g and LOX-1, respectively. MTS, EDU and flow cytometry were performed to evaluate PASMCs proliferation. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were conducted to assess the expression of let-7g, LOX-1, calpain-1,-2,-4, and OCT-1. RESULTS The expression of let-7g was significantly down-regulated in pulmonary arteries of hypoxia-induced PH rats accompanied by pulmonary vascular remodeling, whereas let-7g mimic inhibited hypoxia-induced proliferation of PASMCs and up-regulation of LOX-1 expression. LOX-1 blocking reversed hypoxia-induced down-regulation of let-7g expression. Calpains, protein kinase C and OCT-1 were involved in negative feedback regulation between let-7g and LOX-1. CONCLUSION Negative feedback regulation between let-7g and LOX-1 mediated hypoxia-induced proliferation of in PASMCs.
Collapse
Affiliation(s)
- Wei-Fang Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Tian-Tian Zhu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, Hunan, 410078, China
| | - You-Wen Xiong
- Jiangxi Center of Medical Device Testing, Nanchang, 330029, China
| | - Ai-Zhen Xiong
- Department of Pharmacy, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xiao-Yue Ge
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, Hunan, 410078, China
| | - Chang-Ping Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, Hunan, 410078, China.
| | - Zheng Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, Hunan, 410078, China.
| |
Collapse
|
102
|
Chun HJ, Bonnet S, Chan SY. Translational Advances in the Field of Pulmonary Hypertension. Translating MicroRNA Biology in Pulmonary Hypertension. It Will Take More Than "miR" Words. Am J Respir Crit Care Med 2017; 195:167-178. [PMID: 27648944 PMCID: PMC5394787 DOI: 10.1164/rccm.201604-0886pp] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/10/2016] [Indexed: 12/17/2022] Open
Affiliation(s)
- Hyung J. Chun
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Quebec Heart and Lung Institute Research Centre, University of Laval, Quebec City, Quebec, Canada; and
| | - Stephen Y. Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
103
|
Xiao JH, Hao QY, Wang K, Paul J, Wang YX. Emerging Role of MicroRNAs and Long Noncoding RNAs in Healthy and Diseased Lung. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:343-359. [DOI: 10.1007/978-3-319-63245-2_22] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
104
|
Zhang WF, Xiong YW, Zhu TT, Xiong AZ, Bao HH, Cheng XS. MicroRNA let-7g inhibited hypoxia-induced proliferation of PASMCs via G 0/G 1 cell cycle arrest by targeting c-myc. Life Sci 2016; 170:9-15. [PMID: 27889560 DOI: 10.1016/j.lfs.2016.11.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/17/2016] [Accepted: 11/22/2016] [Indexed: 01/06/2023]
Abstract
AIMS Pulmonary hypertension (PH) is a proliferative disorder characterized by enhanced proliferation and suppressed apoptosis of intrapulmonary vascular smooth muscle cells. Recently, network-based bioinformatics have identified let-7 family, a tumor suppressive microRNA, regulate multiple interacting targets relevant to PH. However, the role of let-7 in vascular homeostasis in PH remains unknown. Thus, we wanted to investigate the role of let-7 in hypoxia-induced PASMCs proliferation and the underlying mechanism in hypoxic pulmonary hypertension (HPH). MAIN METHODS The male Sprague-Dawley (SD) rats were exposed to hypoxia (10% O2) for 21days to induce HPH. The expression of let-7 was determined by quantitative real-time polymerase chain reaction (qRT-PCR) and in situ hybridization. Primary rat PASMCs were exposed to hypoxia (3% O2). MTS and EDU were performed to evaluate PASMCs proliferation. The mRNA and protein expression of c-myc, Bmi-1 and p16 were determined by qRT-PCR and Western blotting, respectively. The functions of let-7g on PASMCs proliferation, c-myc, Bmi-1 and p16 expression were assessed by let-7g mimic and inhibitor transfection. KEY FINDINGS Among let-7 family members, only let-7b and let-7g were significantly down-regulated in remodeled pulmonary artery in HPH rats. Furthermore, only let-7g level was decreased in hypoxic PASMCs. Either hypoxia or let-7g inhibitor stimulated proliferation of PASMCs, let-7g mimic inhibited hypoxia-induced PASMCs proliferation. C-myc was the target of let-7g in PASMCs. Transfect of let-7g mimic inhibited hypoxia-induced c-myc, Bmi-1 up-regulation and p16 down-regulation, which ultimately controls cell cycle progression. SIGNIFICANCE Loss of inhibition on c-myc-Bmi-1-p16 signaling pathway by let-7g may lead to PASMCs proliferation and vascular remodeling in HPH.
Collapse
Affiliation(s)
- Wei-Fang Zhang
- Department of Pharmacy/Cardiovascular medicine, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - You-Wen Xiong
- Jiangxi Supervision and Inspection Center for Medical Devices, Nanchang 330029, China
| | - Tian-Tian Zhu
- Department of Pharmacology, School of Pharmaceutical Science, Central South University, Changsha 410078, China
| | - Ai-Zhen Xiong
- Department of Pharmacy/Cardiovascular medicine, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Hui-Hui Bao
- Department of Pharmacy/Cardiovascular medicine, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiao-Shu Cheng
- Department of Pharmacy/Cardiovascular medicine, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
105
|
Abstract
PURPOSE OF REVIEW Noncoding RNAs (ncRNAs), such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) are becoming fundamentally important in the pathophysiology relating to injury-induced vascular remodelling. We highlight recent studies that demonstrate the involvement of ncRNAs in vein graft disease, in in-stent restenosis and in pulmonary arterial hypertension, with a particular focus on endothelial cell and vascular smooth muscle cell function. We also briefly discuss the emerging role of exosomal-derived ncRNAs and how this mechanism impacts on vascular function. RECENT FINDINGS ncRNAs have been described as novel regulators in the pathophysiology of vascular injury, inflammation, and vessel wall remodelling. In particular, several studies have demonstrated that manipulation of miRNAs can reduce the burden of pathological vascular remodelling. Such studies have also shown that exosomal miRNA-mediated, cell-to-cell communication between endothelial cells and vascular smooth muscle cells is critical in the disease process. In addition to miRNAs, lncRNAs are emerging as regulators of vascular function in health and disease. Although lncRNAs are complex in both their sheer numbers and mechanisms of action, identifying their contribution to vascular disease is essential. SUMMARY Given the important roles of ncRNAs in vascular injury and remodelling together will their capacity for cell-to-cell communication, manipulating ncRNA might provide novel therapeutic interventions.
Collapse
Affiliation(s)
- Lin Deng
- aBHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow bCentre for Cardiovascular Science, Queen's Medical Research Institute, BHF/University of Edinburgh, Edinburgh, UK
| | | | | |
Collapse
|
106
|
Maron BA, Leopold JA. Systems biology: An emerging strategy for discovering novel pathogenetic mechanisms that promote cardiovascular disease. Glob Cardiol Sci Pract 2016; 2016:e201627. [PMID: 29043273 PMCID: PMC5642838 DOI: 10.21542/gcsp.2016.27] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Reductionist theory proposes that analyzing complex systems according to their most fundamental components is required for problem resolution, and has served as the cornerstone of scientific methodology for more than four centuries. However, technological gains in the current scientific era now allow for the generation of large datasets that profile the proteomic, genomic, and metabolomic signatures of biological systems across a range of conditions. The accessibility of data on such a vast scale has, in turn, highlighted the limitations of reductionism, which is not conducive to analyses that consider multiple and contemporaneous interactions between intermediates within a pathway or across constructs. Systems biology has emerged as an alternative approach to analyze complex biological systems. This methodology is based on the generation of scale-free networks and, thus, provides a quantitative assessment of relationships between multiple intermediates, such as protein-protein interactions, within and between pathways of interest. In this way, systems biology is well positioned to identify novel targets implicated in the pathogenesis or treatment of diseases. In this review, the historical root and fundamental basis of systems biology, as well as the potential applications of this methodology are discussed with particular emphasis on integration of these concepts to further understanding of cardiovascular disorders such as coronary artery disease and pulmonary hypertension.
Collapse
Affiliation(s)
- Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Cardiology, Boston VA Healthcare System, Boston, MA, USA
| | - Jane A Leopold
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
107
|
Brittain EL, Chan SY. Integration of complex data sources to provide biologic insight into pulmonary vascular disease (2015 Grover Conference Series). Pulm Circ 2016; 6:251-60. [PMID: 27683602 DOI: 10.1086/686995] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The application of complex data sources to pulmonary vascular diseases is an emerging and promising area of investigation. The use of -omics platforms, in silico modeling of gene networks, and linkage of large human cohorts with DNA biobanks are beginning to bear biologic insight into pulmonary hypertension. These approaches to high-throughput molecular phenotyping offer the possibility of discovering new therapeutic targets and identifying variability in response to therapy that can be leveraged to improve clinical care. Optimizing the methods for analyzing complex data sources and accruing large, well-phenotyped human cohorts linked to biologic data remain significant challenges. Here, we discuss two specific types of complex data sources-gene regulatory networks and DNA-linked electronic medical record cohorts-that illustrate the promise, challenges, and current limitations of these approaches to understanding and managing pulmonary vascular disease.
Collapse
Affiliation(s)
- Evan L Brittain
- Division of Cardiovascular Medicine and Vanderbilt Translational and Clinical Cardiovascular Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Stephen Y Chan
- Division of Cardiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA; and Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
108
|
Sharma S, Ruffenach G, Umar S, Motayagheni N, Reddy ST, Eghbali M. Role of oxidized lipids in pulmonary arterial hypertension. Pulm Circ 2016; 6:261-73. [PMID: 27683603 DOI: 10.1086/687293] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial disease characterized by interplay of many cellular, molecular, and genetic events that lead to excessive proliferation of pulmonary cells, including smooth muscle and endothelial cells; inflammation; and extracellular matrix remodeling. Abnormal vascular changes and structural remodeling associated with PAH culminate in vasoconstriction and obstruction of pulmonary arteries, contributing to increased pulmonary vascular resistance, pulmonary hypertension, and right ventricular failure. The complex molecular mechanisms involved in the pathobiology of PAH are the limiting factors in the development of potential therapeutic interventions for PAH. Over the years, our group and others have demonstrated the critical implication of lipids in the pathogenesis of PAH. This review specifically focuses on the current understanding of the role of oxidized lipids, lipid metabolism, peroxidation, and oxidative stress in the progression of PAH. This review also discusses the relevance of apolipoprotein A-I mimetic peptides and microRNA-193, which are known to regulate the levels of oxidized lipids, as potential therapeutics in PAH.
Collapse
Affiliation(s)
- Salil Sharma
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Grégoire Ruffenach
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Soban Umar
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Negar Motayagheni
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Srinivasa T Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Mansoureh Eghbali
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
109
|
Abstract
PURPOSE OF REVIEW Pulmonary arterial hypertension (PAH) is a rare disease with poor prognosis and no therapeutics. PAH is characterized by severe remodeling of precapillary pulmonary arteries, leading to increased vascular resistance, pulmonary hypertension compensatory right ventricular hypertrophy, then heart failure and death. PAH pathogenesis shares similarities with carcinogenesis such as excessive cell proliferation, apoptosis resistance, metabolic shifts, or phenotypic transition. Although PAH is not a cancer, comparison of analogous mechanisms between PAH and cancer led to the concept of a cancer-like disease to emerge. MicroRNAs (miRNAs) are small noncoding RNAs involved in the regulation of posttranscriptional gene expression. miRNA dysregulations have been reported as promoter of the development of various diseases including cancers. RECENT FINDINGS Recent studies revealed that miRNA dysregulations also occur in PAH pathogenesis. In PAH, different miRNAs have been implicated to be the main features of PAH pathophysiology (in pulmonary inflammation, vascular remodeling, angiogenesis, and right heart hypertrophy). SUMMARY The review summarizes the implication of miRNA dysregulation in PAH development and discusses the similarities and differences with those observed in cancers.
Collapse
|
110
|
Abstract
Transforming growth factor β (TGF-β) family members signal via heterotetrameric complexes of type I and type II dual specificity kinase receptors. The activation and stability of the receptors are controlled by posttranslational modifications, such as phosphorylation, ubiquitylation, sumoylation, and neddylation, as well as by interaction with other proteins at the cell surface and in the cytoplasm. Activation of TGF-β receptors induces signaling via formation of Smad complexes that are translocated to the nucleus where they act as transcription factors, as well as via non-Smad pathways, including the Erk1/2, JNK and p38 MAP kinase pathways, and the Src tyrosine kinase, phosphatidylinositol 3'-kinase, and Rho GTPases.
Collapse
Affiliation(s)
- Carl-Henrik Heldin
- Ludwig Institute for Cancer Research Ltd., Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Aristidis Moustakas
- Ludwig Institute for Cancer Research Ltd., Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23 Uppsala, Sweden
| |
Collapse
|
111
|
Abstract
The cardiovascular research and clinical communities are ideally positioned to address the epidemic of noncommunicable causes of death, as well as advance our understanding of human health and disease, through the development and implementation of precision medicine. New tools will be needed for describing the cardiovascular health status of individuals and populations, including 'omic' data, exposome and social determinants of health, the microbiome, behaviours and motivations, patient-generated data, and the array of data in electronic medical records. Cardiovascular specialists can build on their experience and use precision medicine to facilitate discovery science and improve the efficiency of clinical research, with the goal of providing more precise information to improve the health of individuals and populations. Overcoming the barriers to implementing precision medicine will require addressing a range of technical and sociopolitical issues. Health care under precision medicine will become a more integrated, dynamic system, in which patients are no longer a passive entity on whom measurements are made, but instead are central stakeholders who contribute data and participate actively in shared decision-making. Many traditionally defined diseases have common mechanisms; therefore, elimination of a siloed approach to medicine will ultimately pave the path to the creation of a universal precision medicine environment.
Collapse
Affiliation(s)
- Elliott M Antman
- Brigham and Women's Hospital, TIMI Study Group, 350 Longwood Avenue, Office Level One, Boston, Massachusetts 02115, USA
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, Massachusetts 02115, USA
| |
Collapse
|
112
|
Sheng WZ, Chen YS, Tu CT, He J, Zhang B, Gao WD. MicroRNA-21 promotes phosphatase gene and protein kinase B/phosphatidylinositol 3-kinase expression in colorectal cancer. World J Gastroenterol 2016; 22:5532-5539. [PMID: 27350731 PMCID: PMC4917613 DOI: 10.3748/wjg.v22.i24.5532] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/11/2016] [Accepted: 05/04/2016] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the regulatory mechanism of the target gene of microRNA-21 (miR-21), phosphatase gene (PTEN), and its downstream proteins, protein kinase B (AKT) and phosphatidylinositol 3-kinase (PI3K), in colorectal cancer (CRC) cells.
METHODS: Quantitative real-time PCR (qRT-PCR) and Western blot were used to detect the expression levels of miR-21 and PTEN in HCT116, HT29, Colo32 and SW480 CRC cell lines. Also, the expression levels of PTEN mRNA and its downstream proteins AKT and PI3K in HCT116 cells after downregulating miR-21 were investigated.
RESULTS: Comparing the miR-21 expression in CRC cells, the expression levels of miR-21 were highest in HCT116 cells, and the expression levels of miR-21 were lowest in SW480 cells. In comparing miR-21 and PTEN expression in CRC cells, we found that the protein expression levels of miR-21 and PTEN were inversely correlated (P < 0.05); when miR-21 expression was reduced, mRNA expression levels of PTEN did not significantly change (P > 0.05), but the expression levels of its protein significantly increased (P < 0.05). In comparing the levels of PTEN protein and downstream AKT and PI3K in HCT116 cells after downregulation of miR-21 expression, the levels of AKT and PI3K protein expression significantly decreased (P < 0.05).
CONCLUSION: PTEN is one of the direct target genes of miR-21. Thus, phosphatase gene and its downstream AKT and PI3K expression levels can be regulated by regulating the expression levels of miR-21, which in turn regulates the development of CRC.
Collapse
|
113
|
MicroRNA signature of end-stage idiopathic pulmonary arterial hypertension: clinical correlations and regulation of WNT signaling. J Mol Med (Berl) 2016; 94:849-51. [DOI: 10.1007/s00109-016-1431-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
114
|
Rothman AMK, Arnold ND, Pickworth JA, Iremonger J, Ciuclan L, Allen RMH, Guth-Gundel S, Southwood M, Morrell NW, Thomas M, Francis SE, Rowlands DJ, Lawrie A. MicroRNA-140-5p and SMURF1 regulate pulmonary arterial hypertension. J Clin Invest 2016; 126:2495-508. [PMID: 27214554 DOI: 10.1172/jci83361] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 03/31/2016] [Indexed: 12/21/2022] Open
Abstract
Loss of the growth-suppressive effects of bone morphogenetic protein (BMP) signaling has been demonstrated to promote pulmonary arterial endothelial cell dysfunction and induce pulmonary arterial smooth muscle cell (PASMC) proliferation, leading to the development of pulmonary arterial hypertension (PAH). MicroRNAs (miRs) mediate higher order regulation of cellular function through coordinated modulation of mRNA targets; however, miR expression is altered by disease development and drug therapy. Here, we examined treatment-naive patients and experimental models of PAH and identified a reduction in the levels of miR-140-5p. Inhibition of miR-140-5p promoted PASMC proliferation and migration in vitro. In rat models of PAH, nebulized delivery of miR-140-5p mimic prevented the development of PAH and attenuated the progression of established PAH. Network and pathway analysis identified SMAD-specific E3 ubiquitin protein ligase 1 (SMURF1) as a key miR-140-5p target and regulator of BMP signaling. Evaluation of human tissue revealed that SMURF1 is increased in patients with PAH. miR-140-5p mimic or SMURF1 knockdown in PASMCs altered BMP signaling, further supporting these factors as regulators of BMP signaling. Finally, Smurf1 deletion protected mice from PAH, demonstrating a critical role in disease development. Together, these studies identify both miR-140-5p and SMURF1 as key regulators of disease pathology and as potential therapeutic targets for the treatment of PAH.
Collapse
|
115
|
Frantz RP. Pulmonary arterial hypertension or left heart disease with pulmonary hypertension? Toward noninvasive clarity, but time for a new paradigm. Eur Respir J 2016; 46:299-302. [PMID: 26232475 DOI: 10.1183/13993003.00456-2015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
116
|
Leopold JA, Maron BA. Molecular Mechanisms of Pulmonary Vascular Remodeling in Pulmonary Arterial Hypertension. Int J Mol Sci 2016; 17:ijms17050761. [PMID: 27213345 PMCID: PMC4881582 DOI: 10.3390/ijms17050761] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/01/2016] [Accepted: 04/08/2016] [Indexed: 01/28/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease that is precipitated by hypertrophic pulmonary vascular remodeling of distal arterioles to increase pulmonary artery pressure and pulmonary vascular resistance in the absence of left heart, lung parenchymal, or thromboembolic disease. Despite available medical therapy, pulmonary artery remodeling and its attendant hemodynamic consequences result in right ventricular dysfunction, failure, and early death. To limit morbidity and mortality, attention has focused on identifying the cellular and molecular mechanisms underlying aberrant pulmonary artery remodeling to identify pathways for intervention. While there is a well-recognized heritable genetic component to PAH, there is also evidence of other genetic perturbations, including pulmonary vascular cell DNA damage, activation of the DNA damage response, and variations in microRNA expression. These findings likely contribute, in part, to dysregulation of proliferation and apoptosis signaling pathways akin to what is observed in cancer; changes in cellular metabolism, metabolic flux, and mitochondrial function; and endothelial-to-mesenchymal transition as key signaling pathways that promote pulmonary vascular remodeling. This review will highlight recent advances in the field with an emphasis on the aforementioned molecular mechanisms as contributors to the pulmonary vascular disease pathophenotype.
Collapse
Affiliation(s)
- Jane A Leopold
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Bradley A Maron
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
- Division of Cardiology, Veterans Affairs Boston Healthcare System, Boston, MA 02132, USA.
| |
Collapse
|
117
|
Identifying microRNAs targeting Wnt/β-catenin pathway in end-stage idiopathic pulmonary arterial hypertension. J Mol Med (Berl) 2016; 94:875-85. [PMID: 27188753 DOI: 10.1007/s00109-016-1426-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 04/28/2016] [Accepted: 05/10/2016] [Indexed: 12/17/2022]
Abstract
UNLABELLED MicroRNAs (miRNAs) play important roles in the pathogenesis of pulmonary arterial hypertension (PAH). However, the pathways targeted by miRNAs in PAH have not been systematically investigated. We aim to identify dysregulated miRNAs for patients with idiopathic PAH (IPAH). miRNA profiling was performed on lung tissue total RNA from eight IPAH patients and eight control subjects. Real-time quantitative RT-PCR (qRT-PCR) was used for validation of miRNA and mRNA expression levels in 14 IPAH patients and 14 control subjects. Pathway enrichment analysis showed that Wnt/β-catenin signaling is among the top PAH-related pathways enriched in target genes of dysregulated miRNAs. We confirmed the significant increased expression levels of five miRNAs (let-7a-5p, miR-26b-5p, miR-27b-3p, miR-199a-3p and miR-656) targeting major PAH-related pathways. Moreover, qRT-PCR validation of Wnt/β-catenin pathway activation indicated multiple genes including receptors (FZD4, FZD5), core molecule (CTNNB1), and downstream targets (CCND1, VEGFA, and AXIN2) were significantly upregulated. The expression level of miR-199b-5p was positively correlated with patients' hemodynamics (PVR: r = 0.522, p = 0.038) and pulmonary vascular remodeling (muscularization: r = 0.540, p = 0.021). We confirmed overexpression of miR-199b-5p in hypoxic pulmonary arterial endothelial cells that negatively regulates GSK3B expression. In summary, miRNAs influence the pathogenesis of PAH by regulating major PAH-related pathways including Wnt/β-catenin in end-stage IPAH. KEY MESSAGE It is the first miRNA profiling study in lung tissue from end-stage idiopathic PAH. We identified dysregulated miRNAs and major pathways (e.g., Wnt signaling) in IPAH. Levels of miRNA expression were correlated with hemodynamics and pathological changes. We observed aberrant expression of target genes in the Wnt/β-catenin pathway. miRNAs influence the pathogenesis of PAH by regulating major PAH-related pathways.
Collapse
|
118
|
Mohsenin V. The emerging role of microRNAs in hypoxia-induced pulmonary hypertension. Sleep Breath 2016; 20:1059-67. [DOI: 10.1007/s11325-016-1351-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/11/2016] [Accepted: 04/19/2016] [Indexed: 11/30/2022]
|
119
|
MicroRNA-223 Attenuates Hypoxia-induced Vascular Remodeling by Targeting RhoB/MLC2 in Pulmonary Arterial Smooth Muscle Cells. Sci Rep 2016; 6:24900. [PMID: 27121304 PMCID: PMC4848472 DOI: 10.1038/srep24900] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/06/2016] [Indexed: 01/12/2023] Open
Abstract
There is growing evidence that microRNAs are implicated in pulmonary arterial hypertension (PAH), but underlying mechanisms remain elusive. Here, we identified that miR-223 was significantly downregulated in chronically hypoxic mouse and rat lungs, as well as in pulmonary artery and pulmonary artery smooth muscle cells (PASMC) exposed to hypoxia. Knockdown of miR-223 increased PASMC proliferation. In contrast, miR-223 overexpression abrogated cell proliferation, migration and stress fiber formation. Administering miR-223 agomir in vivo antagonized hypoxia-induced increase in pulmonary artery pressure and distal arteriole muscularization. RhoB, which was increased by hypoxia, was identified as one of the targets of miR-223. Overexpressed miR-223 suppressed RhoB and inhibited the consequent phosphorylation of myosin phosphatase target subunit (MYPT1) and the expression of myosin light chain of myosin II (MLC2), which was identified as another target of miR-223. Furthermore, serum miR-223 levels were decreased in female patients with PAH associated with congenital heart disease. Our study provides the first evidence that miR-223 can regulate PASMC proliferation, migration, and actomyosin reorganization through its novel targets, RhoB and MLC2, resulting in vascular remodeling and the development of PAH. It also highlights miR-223 as a potential circulating biomarker and a small molecule drug for diagnosis and treatment of PAH.
Collapse
|
120
|
Zhu Z, Fang Z, Hu X, Zhou S. MicroRNAs and mesenchymal stem cells: hope for pulmonary hypertension. Braz J Cardiovasc Surg 2016; 30:380-5. [PMID: 26313730 PMCID: PMC4541786 DOI: 10.5935/1678-9741.20150033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/12/2015] [Indexed: 12/21/2022] Open
Abstract
Pulmonary hypertension is a devastating and refractory disease and there is no cure
for this disease. Recently, microRNAs and mesenchymal stem cells emerged as novel
methods to treat pulmonary hypertension. More than 20 kinds of microRNAs may
participate in the process of pulmonary hypertension. It seems microRNAs or
mesenchymal stem cells can ameliorate some symptoms of pulmonary hypertension in
animals and even improve heart and lung function during pulmonary hypertension.
Nevertheless, the relationship between mesenchymal stem cells, microRNAs and
pulmonary hypertension is not clear. And the mechanisms underlying their function
still need to be investigated. In this study we review the recent findings in
mesenchymal stem cells - and microRNAs-based pulmonary hypertension treatment,
focusing on the potential role of microRNAs regulated mesenchymal stem cells in
pulmonary hypertension and the role of exosomes between mesenchymal stem cells and
pulmonary hypertension.
Collapse
Affiliation(s)
- Zhaowei Zhu
- The Second Xiangya Hospital, Central South University, Huan Province, CN
| | - Zhenfei Fang
- Department of Cardiology, Second Xiangya Hospital, Central South University, Huan Province, CN
| | - Xinqun Hu
- Department of Cardiology, Second Xiangya Hospital, Central South University, Huan Province, CN
| | - Shenghua Zhou
- The Second Xiangya Hospital, Central South University, Huan Province, CN
| |
Collapse
|
121
|
Aliotta JM, Pereira M, Wen S, Dooner MS, Del Tatto M, Papa E, Goldberg LR, Baird GL, Ventetuolo CE, Quesenberry PJ, Klinger JR. Exosomes induce and reverse monocrotaline-induced pulmonary hypertension in mice. Cardiovasc Res 2016; 110:319-30. [PMID: 26980205 DOI: 10.1093/cvr/cvw054] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 03/07/2016] [Indexed: 12/13/2022] Open
Abstract
AIMS Extracellular vesicles (EVs) from mice with monocrotaline (MCT)-induced pulmonary hypertension (PH) induce PH in healthy mice, and the exosomes (EXO) fraction of EVs from mesenchymal stem cells (MSCs) can blunt the development of hypoxic PH. We sought to determine whether the EXO fraction of EVs is responsible for modulating pulmonary vascular responses and whether differences in EXO-miR content explains the differential effects of EXOs from MSCs and mice with MCT-PH. METHODS AND RESULTS Plasma, lung EVs from MCT-PH, and control mice were divided into EXO (exosome), microvesicle (MV) fractions and injected into healthy mice. EVs from MSCs were divided into EXO, MV fractions and injected into MCT-treated mice. PH was assessed by right ventricle-to-left ventricle + septum (RV/LV + S) ratio and pulmonary arterial wall thickness-to-diameter (WT/D) ratio. miR microarray analyses were also performed on all EXO populations. EXOs but not MVs from MCT-injured mice increased RV/LV + S, WT/D ratios in healthy mice. MSC-EXOs prevented any increase in RV/LV + S, WT/D ratios when given at the time of MCT injection and reversed the increase in these ratios when given after MCT administration. EXOs from MCT-injured mice and patients with idiopathic pulmonary arterial hypertension (IPAH) contained increased levels of miRs-19b,-20a,-20b, and -145, whereas miRs isolated from MSC-EXOs had increased levels of anti-inflammatory, anti-proliferative miRs including miRs-34a,-122,-124, and -127. CONCLUSION These findings suggest that circulating or MSC-EXOs may modulate pulmonary hypertensive effects based on their miR cargo. The ability of MSC-EXOs to reverse MCT-PH offers a promising potential target for new PAH therapies.
Collapse
Affiliation(s)
- Jason M Aliotta
- Department of Medicine, Division of Hematology/Oncology, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Mandy Pereira
- Department of Medicine, Division of Hematology/Oncology, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| | - Sicheng Wen
- Department of Medicine, Division of Hematology/Oncology, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| | - Mark S Dooner
- Department of Medicine, Division of Hematology/Oncology, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| | - Michael Del Tatto
- Department of Medicine, Division of Hematology/Oncology, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| | - Elaine Papa
- Department of Medicine, Division of Hematology/Oncology, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| | - Laura R Goldberg
- Department of Medicine, Division of Hematology/Oncology, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| | - Grayson L Baird
- Lifespan Biostatistics Core, Rhode Island Hospital, Providence, RI 02903, USA
| | - Corey E Ventetuolo
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Peter J Quesenberry
- Department of Medicine, Division of Hematology/Oncology, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| | - James R Klinger
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| |
Collapse
|
122
|
Samanta S, Balasubramanian S, Rajasingh S, Patel U, Dhanasekaran A, Dawn B, Rajasingh J. MicroRNA: A new therapeutic strategy for cardiovascular diseases. Trends Cardiovasc Med 2016; 26:407-19. [PMID: 27013138 DOI: 10.1016/j.tcm.2016.02.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/13/2016] [Accepted: 02/17/2016] [Indexed: 12/21/2022]
Abstract
Myocardial infarction, atherosclerosis, and hypertension are the most common heart-related diseases that affect both the heart and the blood vessels. Multiple independent risk factors have been shown to be responsible for cardiovascular diseases. The combination of a healthy diet, exercise, and smoking cessation keeps these risk factors in check and helps maintain homeostasis. The dynamic monolayer endothelial cell integrity and cell-cell communication are the fundamental mechanisms in maintaining homeostasis. Recently, it has been revealed that small noncoding RNAs (ncRNAs) play a critical role in regulation of genes involved in either posttranscriptional or pretranslational modifications. They also control diverse biological functions like development, differentiation, growth, and metabolism. Among ncRNAs, the short interfering RNAs (siRNAs), and microRNAs (miRNAs) have been extensively studied, but their specific functions remain largely unknown. In recent years, miRNAs are efficiently studied as one of the important candidates for involvement in most biological processes and have been implicated in many human diseases. Thus, the identification and the respective targets of miRNAs may provide novel molecular insight and new therapeutic strategies to treat diseases. This review summarizes the recent developments and insight on the role of miRNAs in cardiovascular disease prognosis, diagnostic and clinical applications.
Collapse
Affiliation(s)
- Saheli Samanta
- Department of Internal Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS
| | - Sathyamoorthy Balasubramanian
- Department of Internal Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS; Centre for Biotechnology, Anna University, Chennai, Tamil Nadu, India
| | - Sheeja Rajasingh
- Department of Internal Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS
| | - Urmi Patel
- Department of Internal Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS
| | | | - Buddhadeb Dawn
- Department of Internal Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS
| | - Johnson Rajasingh
- Department of Internal Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS; Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS.
| |
Collapse
|
123
|
Grunig G, Baghdassarian A, Park SH, Pylawka S, Bleck B, Reibman J, Berman-Rosenzweig E, Durmus N. Challenges and Current Efforts in the Development of Biomarkers for Chronic Inflammatory and Remodeling Conditions of the Lungs. Biomark Insights 2016; 10:59-72. [PMID: 26917944 PMCID: PMC4756863 DOI: 10.4137/bmi.s29514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/14/2015] [Accepted: 10/18/2015] [Indexed: 02/06/2023] Open
Abstract
This review discusses biomarkers that are being researched for their usefulness to phenotype chronic inflammatory lung diseases that cause remodeling of the lung's architecture. The review focuses on asthma, chronic obstructive pulmonary disease (COPD), and pulmonary hypertension. Bio-markers of environmental exposure and specific classes of biomarkers (noncoding RNA, metabolism, vitamin, coagulation, and microbiome related) are also discussed. Examples of biomarkers that are in clinical use, biomarkers that are under development, and biomarkers that are still in the research phase are discussed. We chose to present examples of the research in biomarker development by diseases, because asthma, COPD, and pulmonary hypertension are distinct entities, although they clearly share processes of inflammation and remodeling.
Collapse
Affiliation(s)
- Gabriele Grunig
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA.; Department of Medicine, New York University School of Medicine, New York, NY, USA
| | - Aram Baghdassarian
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
| | - Sung-Hyun Park
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
| | - Serhiy Pylawka
- College of Dental Medicine, Columbia University, New York, NY, USA
| | - Bertram Bleck
- Department of Medicine, New York University School of Medicine, New York, NY, USA
| | - Joan Reibman
- Department of Medicine, New York University School of Medicine, New York, NY, USA
| | | | - Nedim Durmus
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
124
|
Brief Report: Coordinated Modulation of Circulating miR-21 in HIV, HIV-Associated Pulmonary Arterial Hypertension, and HIV/Hepatitis C Virus Coinfection. J Acquir Immune Defic Syndr 2016; 70:236-41. [PMID: 26473639 DOI: 10.1097/qai.0000000000000741] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Dysregulation of microRNA-21 (miR-21) is independently associated with HIV infection, pulmonary arterial hypertension (PAH), and hepatitis C virus (HCV) infection. To assess the expression of miR-21 in these overlapping comorbidities, we measured plasma miR-21 in HIV with and without PAH and then stratified by concomitant HCV infection. MiR-21 was increased in HIV and HIV-PAH versus uninfected subjects, but it did not differ between these groups. HIV/HCV coinfection correlated with even higher miR-21 levels within the HIV-infected population. These data reveal specific regulation of plasma miR-21 in HIV, HIV/HCV coinfection, and PAH and suggest that miR-21 may integrate complex disease-specific signaling in the setting of HIV infection.
Collapse
|
125
|
Gubrij IB, Pangle AK, Pang L, Johnson LG. Reversal of MicroRNA Dysregulation in an Animal Model of Pulmonary Hypertension. PLoS One 2016; 11:e0147827. [PMID: 26815432 PMCID: PMC4731388 DOI: 10.1371/journal.pone.0147827] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 01/08/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Animals models have played an important role in enhancing our understanding of the pathogenesis of pulmonary arterial hypertension (PAH). Dysregulation of the profile of microRNAs (miRNAs) has been demonstrated in human tissues from PAH patients and in animal models. In this study, we measured miRNA levels in the monocrotaline (MCT) rat model of PAH and examined whether blocking a specific dysregulated miRNA not previously reported in this model, attenuated PAH. We also evaluated changes in miRNA expression in lung specimens from MCT PAH rats overexpressing human prostacyclin synthase, which has been shown to attenuate MCT PAH. METHODS Expression levels of a panel of miRNAs were measured in MCT-PAH rats as compared to naïve (saline) control rats. Subsequently, MCT PAH rats were injected with a specific inhibitor (antagomiR) for miR-223 (A223) or a nonspecific control oligonucleotide (A-control) 4 days after MCT administration, then weekly. Three weeks later, RV systolic pressure and RV mass were measured. Total RNA, isolated from the lungs, microdissected pulmonary arteries, and right ventricle, was reverse transcribed and real-time quantitative PCR was performed. MiRNA levels were also measured in RNA isolated from paraffin sections of MCT-PAH rats overexpressing prostacyclin synthase. RESULTS MiRs 17, 21, and 223 were consistently upregulated, whereas miRs 126, 145, 150, 204, 424, and 503 were downregulated in MCT PAH as compared to vehicle control. A223 significantly reduced levels of miR-223 in PA and lungs of MCT PAH rats as compared to levels measured in A-control or control MCT PAH rats, but A223 did not attenuate MCT PAH. Right ventricular mass and right ventricular systolic pressure in rats treated with A223 were not different from values in A-control or MCT PAH rats. In contrast, analysis of total RNA from lung specimens of MCT PAH rats overexpressing human prostacyclin synthase (hPGIS) demonstrated reversal of MCT-induced upregulation of miRs 17, 21, and 223 and an increase in levels of miR-424 and miR-503. Reduction in bone morphogenetic receptor 2 (BMPR2) messenger (m)RNA expression was not altered by A223, whereas human prostacyclin synthase overexpression restored BMPR2 mRNA to levels in MCT PAH to levels measured in naive controls. CONCLUSIONS Inhibition of miR-223 did not attenuate MCT PAH, whereas human prostacyclin synthase overexpression restored miRNA levels in MCT PAH to levels detected in naïve rats. These data may establish a paradigm linking attenuation of PAH to restoration of BMPR2 signaling.
Collapse
Affiliation(s)
- Igor B. Gubrij
- Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, United States of America
- Department of Internal Medicine, Division of Pulmonary and Critical Medicine, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Amanda K. Pangle
- Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, United States of America
- Department of Internal Medicine, Division of Pulmonary and Critical Medicine, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Li Pang
- Department of Pharmacology, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Larry G. Johnson
- Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, United States of America
- Department of Internal Medicine, Division of Pulmonary and Critical Medicine, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| |
Collapse
|
126
|
Reconciling paradigms of abnormal pulmonary blood flow and quasi-malignant cellular alterations in pulmonary arterial hypertension. Vascul Pharmacol 2016; 83:17-25. [PMID: 26804008 DOI: 10.1016/j.vph.2016.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 01/19/2016] [Indexed: 01/29/2023]
Abstract
In pulmonary arterial hypertension (PAH) structural and functional abnormalities of the small lung vessels interact and lead to a progressive increase in pulmonary vascular resistance and right heart failure. A current pathobiological concept characterizes PAH as a 'quasi-malignant' disease focusing on cancer-like alterations in endothelial cells (EC) and the importance of their acquired apoptosis-resistant, hyper-proliferative phenotype in the process of vascular remodeling. While changes in pulmonary blood flow (PBF) have been long-since recognized and linked to the development of PAH, little is known about a possible relationship between an altered PBF and the quasi-malignant cell phenotype in the pulmonary vascular wall. This review summarizes recognized and hypothetical effects of an abnormal PBF on the pulmonary vascular bed and links these to quasi-malignant changes found in the pulmonary endothelium. Here we describe that abnormal PBF does not only trigger a pulmonary vascular cell growth program, but may also maintain the cancer-like phenotype of the endothelium. Consequently, normalization of PBF and EC response to abnormal PBF may represent a treatment strategy in patients with established PAH.
Collapse
|
127
|
Bertero T, Cottrill KA, Annis S, Bhat B, Gochuico BR, Osorio JC, Rosas I, Haley KJ, Corey KE, Chung RT, Nelson Chau B, Chan SY. A YAP/TAZ-miR-130/301 molecular circuit exerts systems-level control of fibrosis in a network of human diseases and physiologic conditions. Sci Rep 2015; 5:18277. [PMID: 26667495 PMCID: PMC4678880 DOI: 10.1038/srep18277] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/08/2015] [Indexed: 01/18/2023] Open
Abstract
The molecular origins of fibrosis affecting multiple tissue beds remain incompletely defined. Previously, we delineated the critical role of the control of extracellular matrix (ECM) stiffening by the mechanosensitive microRNA-130/301 family, as activated by the YAP/TAZ co-transcription factors, in promoting pulmonary hypertension (PH). We hypothesized that similar mechanisms may dictate fibrosis in other tissue beds beyond the pulmonary vasculature. Employing an in silico combination of microRNA target prediction, transcriptomic analysis of 137 human diseases and physiologic states, and advanced gene network modeling, we predicted the microRNA-130/301 family as a master regulator of fibrotic pathways across a cohort of seemingly disparate diseases and conditions. In two such diseases (pulmonary fibrosis and liver fibrosis), inhibition of microRNA-130/301 prevented the induction of ECM modification, YAP/TAZ, and downstream tissue fibrosis. Thus, mechanical forces act through a central feedback circuit between microRNA-130/301 and YAP/TAZ to sustain a common fibrotic phenotype across a network of human physiologic and pathophysiologic states. Such re-conceptualization of interconnections based on shared systems of disease and non-disease gene networks may have broad implications for future convergent diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Thomas Bertero
- Divisions of Cardiovascular and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Katherine A. Cottrill
- Divisions of Cardiovascular and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Sofia Annis
- Divisions of Cardiovascular and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | | | | | - Juan C. Osorio
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Ivan Rosas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Kathleen J. Haley
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Kathleen E. Corey
- Liver Center and Gastrointestinal Division, Massachusetts General Hospital, Boston, MA, USA
| | - Raymond T. Chung
- Liver Center and Gastrointestinal Division, Massachusetts General Hospital, Boston, MA, USA
| | | | - Stephen Y. Chan
- Divisions of Cardiovascular and Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| |
Collapse
|
128
|
Ma K, Zhao Q, Chen W, Zhang H, Li S, Pan X, Chen Q. Human lung microRNA profiling in pulmonary arterial hypertension secondary to congenital heart defect. Pediatr Pulmonol 2015; 50:1214-23. [PMID: 25847058 DOI: 10.1002/ppul.23181] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 12/28/2014] [Accepted: 02/10/2015] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Although several microRNAs were reported to play essential roles in pulmonary artery hypertension due to hypoxia or monocrotaline, their potential role in pulmonary arterial hypertension secondary to congenital heart disease is largely unknown. This study aimed to indentify microRNAs implicated in pulmonary arterial hypertension secondary to congenital heart disease in children. METHODS Using microRNAs microarray, we profiled the microRNAs in the lung specimen from 12 congenital heart disease patients, (6 with pulmonary arterial hypertension and the others without). We validated the microRNAs expression using RT-PCR experiments. Then, we predicted the target genes of the promising microRNAs by bioinformatical analysis and verified its regulating role by luciferase assay and western blot experiments. RESULTS All the 12 patients were uneventfully recovered from cardiac surgery. Comparing to the non-pulmonary arterial hypertension lung tissue, 62 microRNAs were significantly up-regulated and 12 were significantly de-regulated in the pulmonary arterial hypertension lung tissue. Among them 27 microRNAs reached P values ≤ 0.05, we validated the up-regulation of microRNA-27b by RT-PCR experiments and found the expression level of microRNA-27b was correlated with preoperative mean pulmonary arterial pressure. In vitro, overexpression of microRNA-27b decreased the protein expression of NOTCH1 and significantly reduced luciferase activity. CONCLUSIONS The current study revealed for the first time that microRNAs may be important regulators in pulmonary arterial hypertension secondary to congenital heart disease, and demonstrated the correlation between microRNA-27b and pulmonary arterial hypertension with the implication of NOTCH1.
Collapse
Affiliation(s)
- Kai Ma
- Department of Pediatric Cardiac Surgery, National Center for Cardiovascular Disease and Fuwai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Qian Zhao
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease and Fuwai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Weidan Chen
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China
| | - Hao Zhang
- Department of Pediatric Cardiac Surgery, National Center for Cardiovascular Disease and Fuwai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Shoujun Li
- Department of Pediatric Cardiac Surgery, National Center for Cardiovascular Disease and Fuwai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Xiangbin Pan
- Department of Pediatric Cardiac Surgery, National Center for Cardiovascular Disease and Fuwai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| | - Qiuming Chen
- Department of Pediatric Cardiac Surgery, National Center for Cardiovascular Disease and Fuwai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China
| |
Collapse
|
129
|
Bertero T, Cottrill KA, Lu Y, Haeger CM, Dieffenbach P, Annis S, Hale A, Bhat B, Kaimal V, Zhang YY, Graham BB, Kumar R, Saggar R, Saggar R, Wallace WD, Ross DJ, Black SM, Fratz S, Fineman JR, Vargas SO, Haley KJ, Waxman AB, Chau BN, Fredenburgh LE, Chan SY. Matrix Remodeling Promotes Pulmonary Hypertension through Feedback Mechanoactivation of the YAP/TAZ-miR-130/301 Circuit. Cell Rep 2015; 13:1016-32. [PMID: 26565914 PMCID: PMC4644508 DOI: 10.1016/j.celrep.2015.09.049] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 08/07/2015] [Accepted: 09/17/2015] [Indexed: 12/21/2022] Open
Abstract
Pulmonary hypertension (PH) is a deadly vascular disease with enigmatic molecular origins. We found that vascular extracellular matrix (ECM) remodeling and stiffening are early and pervasive processes that promote PH. In multiple pulmonary vascular cell types, such ECM stiffening induced the microRNA-130/301 family via activation of the co-transcription factors YAP and TAZ. MicroRNA-130/301 controlled a PPAR?-APOE-LRP8 axis, promoting collagen deposition and LOX-dependent remodeling and further upregulating YAP/TAZ via a mechanoactive feedback loop. In turn, ECM remodeling controlled pulmonary vascular cell crosstalk via such mechanotransduction, modulation of secreted vasoactive effectors, and regulation of associated microRNA pathways. In vivo, pharmacologic inhibition of microRNA-130/301, APOE, or LOX activity ameliorated ECM remodeling and PH. Thus, ECM remodeling, as controlled by the YAP/TAZ-miR-130/301 feedback circuit, is an early PH trigger and offers combinatorial therapeutic targets for this devastating disease.
Collapse
Affiliation(s)
- Thomas Bertero
- Divisions of Cardiovascular and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Katherine A Cottrill
- Divisions of Cardiovascular and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yu Lu
- Divisions of Cardiovascular and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christina M Haeger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Paul Dieffenbach
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sofia Annis
- Divisions of Cardiovascular and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew Hale
- Divisions of Cardiovascular and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Ying-Yi Zhang
- Divisions of Cardiovascular and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Brian B Graham
- Program in Translational Lung Research, University of Colorado, Denver, Aurora, CO 80045, USA
| | - Rahul Kumar
- Program in Translational Lung Research, University of Colorado, Denver, Aurora, CO 80045, USA
| | - Rajan Saggar
- Departments of Medicine and Pathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rajeev Saggar
- Department of Medicine, University of Arizona, Phoenix, AZ 85006, USA
| | - W Dean Wallace
- Departments of Medicine and Pathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - David J Ross
- Departments of Medicine and Pathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stephen M Black
- Department of Medicine, University of Arizona, Tuscon, AZ 85724, USA
| | - Sohrab Fratz
- Department of Pediatric Cardiology and Congenital Heart Disease, DeutschesHerzzentrum München, Klinik an der Technischen Universität München, 80636 Munich, Germany
| | - Jeffrey R Fineman
- Department of Pediatrics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94131, USA
| | - Sara O Vargas
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Kathleen J Haley
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Aaron B Waxman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Laura E Fredenburgh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Stephen Y Chan
- Divisions of Cardiovascular and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
130
|
Rhodes CJ, Im H, Cao A, Hennigs JK, Wang L, Sa S, Chen PI, Nickel NP, Miyagawa K, Hopper RK, Tojais NF, Li CG, Gu M, Spiekerkoetter E, Xian Z, Chen R, Zhao M, Kaschwich M, Del Rosario PA, Bernstein D, Zamanian RT, Wu JC, Snyder MP, Rabinovitch M. RNA Sequencing Analysis Detection of a Novel Pathway of Endothelial Dysfunction in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2015; 192:356-66. [PMID: 26030479 DOI: 10.1164/rccm.201408-1528oc] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Pulmonary arterial hypertension is characterized by endothelial dysregulation, but global changes in gene expression have not been related to perturbations in function. OBJECTIVES RNA sequencing was used to discriminate changes in transcriptomes of endothelial cells cultured from lungs of patients with idiopathic pulmonary arterial hypertension versus control subjects and to assess the functional significance of major differentially expressed transcripts. METHODS The endothelial transcriptomes from the lungs of seven control subjects and six patients with idiopathic pulmonary arterial hypertension were analyzed. Differentially expressed genes were related to bone morphogenetic protein type 2 receptor (BMPR2) signaling. Those down-regulated were assessed for function in cultured cells and in a transgenic mouse. MEASUREMENTS AND MAIN RESULTS Fold differences in 10 genes were significant (P < 0.05), four increased and six decreased in patients versus control subjects. No patient was mutant for BMPR2. However, knockdown of BMPR2 by siRNA in control pulmonary arterial endothelial cells recapitulated 6 of 10 patient-related gene changes, including decreased collagen IV (COL4A1, COL4A2) and ephrinA1 (EFNA1). Reduction of BMPR2-regulated transcripts was related to decreased β-catenin. Reducing COL4A1, COL4A2, and EFNA1 by siRNA inhibited pulmonary endothelial adhesion, migration, and tube formation. In mice null for the EFNA1 receptor, EphA2, versus control animals, vascular endothelial growth factor receptor blockade and hypoxia caused more severe pulmonary hypertension, judged by elevated right ventricular systolic pressure, right ventricular hypertrophy, and loss of small arteries. CONCLUSIONS The novel relationship between BMPR2 dysfunction and reduced expression of endothelial COL4 and EFNA1 may underlie vulnerability to injury in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Christopher J Rhodes
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Hogune Im
- 2 Cardiovascular Institute.,4 Department of Genetics, and
| | - Aiqin Cao
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Jan K Hennigs
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Lingli Wang
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Silin Sa
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Pin-I Chen
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Nils P Nickel
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Kazuya Miyagawa
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Rachel K Hopper
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Nancy F Tojais
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Caiyun G Li
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Mingxia Gu
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Edda Spiekerkoetter
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,5 Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Zhaoying Xian
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Rui Chen
- 2 Cardiovascular Institute.,4 Department of Genetics, and
| | - Mingming Zhao
- 2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Mark Kaschwich
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Patricia A Del Rosario
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,5 Department of Medicine, Stanford University School of Medicine, Stanford, California
| | | | - Roham T Zamanian
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,5 Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Joseph C Wu
- 2 Cardiovascular Institute.,5 Department of Medicine, Stanford University School of Medicine, Stanford, California
| | | | - Marlene Rabinovitch
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| |
Collapse
|
131
|
Schlosser K, Taha M, Deng Y, Jiang B, Stewart DJ. Discordant Regulation of microRNA Between Multiple Experimental Models and Human Pulmonary Hypertension. Chest 2015; 148:481-490. [PMID: 25763574 DOI: 10.1378/chest.14-2169] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND The dysregulation of microRNA (miRNA) is known to contribute to the pathobiology of pulmonary arterial hypertension (PAH). However, the relationships between changes in tissue and circulating miRNA levels associated with different animal models and human pulmonary hypertension (PH) have not been defined. METHODS A set of miRNAs that have been causally implicated in PH, including miR-17, -21, -130b, -145, -204, -424, and -503, were measured by reverse transcription-quantitative polymerase chain reaction in the plasma, lung, and right ventricle of three of the most common rodent models of PH: the rat monocrotaline and SU5416 plus chronic hypoxia (SuHx) models and the mouse chronic hypoxia model. Plasma miRNA levels were also evaluated in a cohort of patients with PAH and healthy subjects. RESULTS Several miRNA showed PH model-dependent perturbations in plasma and tissue levels; however, none of these were conserved across all three experimental models. Principle component analysis of miR expression changes in plasma revealed distinct clustering between rodent models, and SuHx-triggered PH showed the greatest similarity to human PAH. Changes in the plasma levels of several miRNA also correlated with changes in tissue expression. In particular, miR-424 was concordantly increased (1.3- to 1.5-fold, P < .05) in the plasma, lung, and right ventricle of hypoxic mice and in the plasma of patients with PAH. CONCLUSIONS miRNAs with established etiologic roles in PH showed context-dependent changes in tissue and circulating levels, which were not consistent across rodent models and human PAH. This suggests different miRNA-dependent mechanisms may contribute to experimental and clinical PH, complicating potential diagnostic and therapeutic applications amenable to these miRNAs.
Collapse
Affiliation(s)
- Kenny Schlosser
- From the Regenerative Medicine Program, Ottawa Hospital Research Institute), Ottawa, ON, Canada
| | - Mohamad Taha
- From the Regenerative Medicine Program, Ottawa Hospital Research Institute), Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Yupu Deng
- From the Regenerative Medicine Program, Ottawa Hospital Research Institute), Ottawa, ON, Canada
| | - Baohua Jiang
- From the Regenerative Medicine Program, Ottawa Hospital Research Institute), Ottawa, ON, Canada
| | - Duncan J Stewart
- From the Regenerative Medicine Program, Ottawa Hospital Research Institute), Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
132
|
Chen X, Talati M, Fessel JP, Hemnes AR, Gladson S, French J, Shay S, Trammell A, Phillips JA, Hamid R, Cogan JD, Dawson EP, Womble KE, Hedges LK, Martinez EG, Wheeler LA, Loyd JE, Majka SJ, West J, Austin ED. Estrogen Metabolite 16α-Hydroxyestrone Exacerbates Bone Morphogenetic Protein Receptor Type II-Associated Pulmonary Arterial Hypertension Through MicroRNA-29-Mediated Modulation of Cellular Metabolism. Circulation 2015; 133:82-97. [PMID: 26487756 DOI: 10.1161/circulationaha.115.016133] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 10/02/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a proliferative disease of the pulmonary vasculature that preferentially affects women. Estrogens such as the metabolite 16α-hydroxyestrone (16αOHE) may contribute to PAH pathogenesis, and alterations in cellular energy metabolism associate with PAH. We hypothesized that 16αOHE promotes heritable PAH (HPAH) via microRNA-29 (miR-29) family upregulation and that antagonism of miR-29 would attenuate pulmonary hypertension in transgenic mouse models of Bmpr2 mutation. METHODS AND RESULTS MicroRNA array profiling of human lung tissue found elevation of microRNAs associated with energy metabolism, including the miR-29 family, among HPAH patients. miR-29 expression was 2-fold higher in Bmpr2 mutant mice lungs at baseline compared with controls and 4 to 8-fold higher in Bmpr2 mice exposed to 16αOHE 1.25 μg/h for 4 weeks. Blot analyses of Bmpr2 mouse lung protein showed significant reductions in peroxisome proliferator-activated receptor-γ and CD36 in those mice exposed to 16αOHE and protein derived from HPAH lungs compared with controls. Bmpr2 mice treated with anti-miR-29 (20-mg/kg injections for 6 weeks) had improvements in hemodynamic profile, histology, and markers of dysregulated energy metabolism compared with controls. Pulmonary artery smooth muscle cells derived from Bmpr2 murine lungs demonstrated mitochondrial abnormalities, which improved with anti-miR-29 transfection in vitro; endothelial-like cells derived from HPAH patient induced pluripotent stem cell lines were similar and improved with anti-miR-29 treatment. CONCLUSIONS 16αOHE promotes the development of HPAH via upregulation of miR-29, which alters molecular and functional indexes of energy metabolism. Antagonism of miR-29 improves in vivo and in vitro features of HPAH and reveals a possible novel therapeutic target.
Collapse
Affiliation(s)
- Xinping Chen
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Megha Talati
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Joshua P Fessel
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Anna R Hemnes
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Santhi Gladson
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Jaketa French
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Sheila Shay
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Aaron Trammell
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - John A Phillips
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Rizwan Hamid
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Joy D Cogan
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Elliott P Dawson
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Kristie E Womble
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Lora K Hedges
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Elizabeth G Martinez
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Lisa A Wheeler
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - James E Loyd
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Susan J Majka
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - James West
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.)
| | - Eric D Austin
- From Departments of Medicine (X.C., M.T., J.P.F., A.R.H., S.G., J.F., S.S., L.A.W., J.E.L., S.J.M., J.W.), Pharmacology (J.P.F.), Pediatrics (J.A.P., R.H., J..C., L.K.H.), and Pathology (E.G.M.), Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Baylor College of Medicine, Houston, TX (A.T.); and Bioventures, Inc, Murfreesboro, TN (E.P.D., K.E.W.).
| |
Collapse
|
133
|
Sharma S, Umar S, Centala A, Eghbali M. Role of miR206 in genistein-induced rescue of pulmonary hypertension in monocrotaline model. J Appl Physiol (1985) 2015; 119:1374-82. [PMID: 26472874 DOI: 10.1152/japplphysiol.00699.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 10/03/2015] [Indexed: 11/22/2022] Open
Abstract
Pulmonary hypertension (PH) is a progressive lung disease associated with proliferation of smooth muscle cells and constriction of lung microvasculature, leading to increased pulmonary arterial pressure, right ventricular failure, and death. We have previously shown that genistein rescues preexisting established PH by significantly improving lung and heart function. (Matori H, Umar S, Nadadur RD, Sharma S, Partow-Navid R, Afkhami M, Amjedi M, Eghbali M. Hypertension 60: 425-430, 2012). Here, we have examined the role of microRNAs (miRs) in the rescue action of genistein in monocrotaline (MCT)-induced PH in rats. Our miR microarray analysis on the lung samples from control, PH, and genistein-rescue group revealed that miR206, which was robustly upregulated to ∼11-fold by PH, was completely normalized to control levels by genistein treatment. Next, we examined whether knockdown of miR206 could reverse preexisting established PH. PH was induced in male rats by 60 mg/kg of MCT, and rats received three intratracheal doses of either miR206 antagomir (10 mg/kg body wt) or scrambled miR control at days 17, 21, and 26. Knockdown of miR206 resulted in significant improvement in the cardiopulmonary function, as right ventricular pressure was significantly reduced to 38.6 ± 3.61 mmHg from 61.2 ± 5.4 mmHg in PH, and right ventricular hypertrophy index was decreased to 0.35 ± 0.04 from 0.59 ± 0.037 in PH. Knockdown of miR206 reversed PH-induced pulmonary vascular remodeling in vivo and was associated with restoration of PH-induced loss of capillaries in the lungs and induction of vascular endothelial growth factor A expression. In conclusion, miR206 antagomir therapy improves cardiopulmonary function and structure and rescues preexisting severe PH in MCT rat model possibly by stimulating angiogenesis in the lung.
Collapse
Affiliation(s)
- Salil Sharma
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and
| | - Soban Umar
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and
| | - Alexander Centala
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and
| | - Mansoureh Eghbali
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California; and Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
134
|
Green DE, Murphy TC, Kang BY, Searles CD, Hart CM. PPARγ Ligands Attenuate Hypoxia-Induced Proliferation in Human Pulmonary Artery Smooth Muscle Cells through Modulation of MicroRNA-21. PLoS One 2015. [PMID: 26208095 PMCID: PMC4514882 DOI: 10.1371/journal.pone.0133391] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pulmonary hypertension (PH) is a progressive and often fatal disorder whose pathogenesis involves pulmonary artery smooth muscle cell (PASMC) proliferation. Although modern PH therapies have significantly improved survival, continued progress rests on the discovery of novel therapies and molecular targets. MicroRNA (miR)-21 has emerged as an important non-coding RNA that contributes to PH pathogenesis by enhancing vascular cell proliferation, however little is known about available therapies that modulate its expression. We previously demonstrated that peroxisome proliferator-activated receptor gamma (PPARγ) agonists attenuated hypoxia-induced HPASMC proliferation, vascular remodeling and PH through pleiotropic actions on multiple targets, including transforming growth factor (TGF)-β1 and phosphatase and tensin homolog deleted on chromosome 10 (PTEN). PTEN is a validated target of miR-21. We therefore hypothesized that antiproliferative effects conferred by PPARγ activation are mediated through inhibition of hypoxia-induced miR-21 expression. Human PASMC monolayers were exposed to hypoxia then treated with the PPARγ agonist, rosiglitazone (RSG,10 μM), or in parallel, C57Bl/6J mice were exposed to hypoxia then treated with RSG. RSG attenuated hypoxic increases in miR-21 expression in vitro and in vivo and abrogated reductions in PTEN and PASMC proliferation. Antiproliferative effects of RSG were lost following siRNA-mediated PTEN depletion. Furthermore, miR-21 mimic decreased PTEN and stimulated PASMC proliferation, whereas miR-21 inhibition increased PTEN and attenuated hypoxia-induced HPASMC proliferation. Collectively, these results demonstrate that PPARγ ligands regulate proliferative responses to hypoxia by preventing hypoxic increases in miR-21 and reductions in PTEN. These findings further clarify molecular mechanisms that support targeting PPARγ to attenuate pathogenic derangements in PH.
Collapse
Affiliation(s)
- David E Green
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center / Emory University, Atlanta, GA, United States of America
| | - Tamara C Murphy
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center / Emory University, Atlanta, GA, United States of America
| | - Bum-Yong Kang
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center / Emory University, Atlanta, GA, United States of America
| | - Charles D Searles
- Department of Medicine, Division of Cardiology, Atlanta Veterans Affairs Medical Center / Emory University, Atlanta, GA, United States of America
| | - C Michael Hart
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs Medical Center / Emory University, Atlanta, GA, United States of America
| |
Collapse
|
135
|
Kurakula K, Goumans MJ, Ten Dijke P. Regulatory RNAs controlling vascular (dys)function by affecting TGF-ß family signalling. EXCLI JOURNAL 2015; 14:832-50. [PMID: 26862319 PMCID: PMC4743484 DOI: 10.17179/excli2015-423] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 06/30/2015] [Indexed: 01/15/2023]
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide. Over the last few years, microRNAs (miRNAs) have emerged as master regulators of gene expression in cardiovascular biology and disease. miRNAs are small endogenous non-coding RNAs that usually bind to 3′ untranslated region (UTR) of their target mRNAs and inhibit mRNA stability or translation of their target genes. miRNAs play a dynamic role in the pathophysiology of many CVDs through their effects on target mRNAs in vascular cells. Recently, numerous miRNAs have been implicated in the regulation of the transforming growth factor-β (TGF-β)/bone morphogenetic protein (BMP) signalling pathway which plays crucial roles in diverse biological processes, and is involved in pathogenesis of many diseases including CVD. This review gives an overview of current literature on the role of miRNAs targeting TGF-β/BMP signalling in vascular cells, including endothelial cells and smooth muscle cells. We also provide insight into how this miRNA-mediated regulation of TGF-β/BMP signalling might be used to harness CVD.
Collapse
Affiliation(s)
- Kondababu Kurakula
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie-Jose Goumans
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
136
|
Mishra A, Mohammad G, Norboo T, Newman JH, Pasha MAQ. Lungs at high-altitude: genomic insights into hypoxic responses. J Appl Physiol (1985) 2015; 119:1-15. [DOI: 10.1152/japplphysiol.00513.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 04/20/2015] [Indexed: 11/22/2022] Open
Abstract
Hypobaric hypoxia at high altitude (HA) results in reduced blood arterial oxygen saturation, perfusion of organs with hypoxemic blood, and direct hypoxia of lung tissues. The pulmonary complications in the cells of the pulmonary arterioles due to hypobaric hypoxia are the basis of the pathophysiological mechanisms of high-altitude pulmonary edema (HAPE). Some populations that have dwelled at HA for thousands of years have evolutionarily adapted to this environmental stress; unadapted populations may react with excessive physiological responses that impair health. Individual variations in response to hypoxia and the mechanisms of HA adaptation provide insight into physiological responses. Adaptive and maladaptive responses include alterations in pathways such as oxygen sensing, hypoxia signaling, K+- and Ca2+-gated channels, redox balance, and the renin-angiotensin-aldosterone system. Physiological imbalances are linked with genetic susceptibilities, and nonhomeostatic responses in gene regulation that occur by small RNAs, histone modification, and DNA methylation predispose susceptible humans to these HA illnesses. Elucidation of the interaction of these factors will lead to a more comprehensive understanding of HA adaptations and maladaptations and will lead to new therapeutics for HA disorders related to hypoxic lungs.
Collapse
Affiliation(s)
- Aastha Mishra
- Department of Genomics and Molecular Medicine, Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
- Department of Biotechnology, University of Pune, Pune, India
| | - Ghulam Mohammad
- Department of Medicine, SNM Hospital, Leh, Ladakh, J&K, India
| | - Tsering Norboo
- Ladakh Institute of Prevention, Leh, Ladakh, J&K, India; and
| | - John H. Newman
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - M. A. Qadar Pasha
- Department of Genomics and Molecular Medicine, Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| |
Collapse
|
137
|
Abstract
Since their initial discovery in the early 1990s, microRNAs have now become the focus of a multitude of lines of investigation ranging from basic biology to translational applications in the clinic. Previously believed to be of no biological relevance, microRNAs regulate processes fundamental to human health and disease. In diseases of the lung, microRNAs have been implicated in developmental programming, as drivers of disease, potential therapeutic targets, and clinical biomarkers; however, several obstacles must be overcome for us to fully realize their potential therapeutic use. Here, we provide for the clinician an overview of microRNA biology in selected diseases of the lung with a focus on their potential clinical application.
Collapse
|
138
|
Luo JY, Zhang Y, Wang L, Huang Y. Regulators and effectors of bone morphogenetic protein signalling in the cardiovascular system. J Physiol 2015; 593:2995-3011. [PMID: 25952563 DOI: 10.1113/jp270207] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/27/2015] [Indexed: 12/22/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) play key roles in the regulation of cell proliferation, differentiation and apoptosis in various tissues and organs, including the cardiovascular system. BMPs signal through both Smad-dependent and -independent cascades to exert a wide spectrum of biological activities. Cardiovascular disorders such as abnormal angiogenesis, atherosclerosis, pulmonary hypertension and cardiac hypertrophy have been linked to aberrant BMP signalling. To correct the dysregulated BMP signalling in cardiovascular pathogenesis, it is essential to get a better understanding of how the regulators and effectors of BMP signalling control cardiovascular function and how the dysregulated BMP signalling contributes to cardiovascular dysfunction. We hence highlight several key regulators of BMP signalling such as extracellular regulators of ligands, mechanical forces, microRNAs and small molecule drugs as well as typical BMP effectors like direct downstream target genes, mitogen-activated protein kinases, reactive oxygen species and microRNAs. The insights into these molecular processes will help target both the regulators and important effectors to reverse BMP-associated cardiovascular pathogenesis.
Collapse
Affiliation(s)
- Jiang-Yun Luo
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yang Zhang
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China.,Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - Li Wang
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yu Huang
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
139
|
White K, Lu Y, Annis S, Hale AE, Chau BN, Dahlman JE, Hemann C, Opotowsky AR, Vargas SO, Rosas I, Perrella MA, Osorio JC, Haley KJ, Graham BB, Kumar R, Saggar R, Saggar R, Wallace WD, Ross DJ, Khan OF, Bader A, Gochuico BR, Matar M, Polach K, Johannessen NM, Prosser HM, Anderson DG, Langer R, Zweier JL, Bindoff LA, Systrom D, Waxman AB, Jin RC, Chan SY. Genetic and hypoxic alterations of the microRNA-210-ISCU1/2 axis promote iron-sulfur deficiency and pulmonary hypertension. EMBO Mol Med 2015; 7:695-713. [PMID: 25825391 PMCID: PMC4459813 DOI: 10.15252/emmm.201404511] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Revised: 02/20/2015] [Accepted: 02/23/2015] [Indexed: 12/03/2022] Open
Abstract
Iron-sulfur (Fe-S) clusters are essential for mitochondrial metabolism, but their regulation in pulmonary hypertension (PH) remains enigmatic. We demonstrate that alterations of the miR-210-ISCU1/2 axis cause Fe-S deficiencies in vivo and promote PH. In pulmonary vascular cells and particularly endothelium, hypoxic induction of miR-210 and repression of the miR-210 targets ISCU1/2 down-regulated Fe-S levels. In mouse and human vascular and endothelial tissue affected by PH, miR-210 was elevated accompanied by decreased ISCU1/2 and Fe-S integrity. In mice, miR-210 repressed ISCU1/2 and promoted PH. Mice deficient in miR-210, via genetic/pharmacologic means or via an endothelial-specific manner, displayed increased ISCU1/2 and were resistant to Fe-S-dependent pathophenotypes and PH. Similar to hypoxia or miR-210 overexpression, ISCU1/2 knockdown also promoted PH. Finally, cardiopulmonary exercise testing of a woman with homozygous ISCU mutations revealed exercise-induced pulmonary vascular dysfunction. Thus, driven by acquired (hypoxia) or genetic causes, the miR-210-ISCU1/2 regulatory axis is a pathogenic lynchpin causing Fe-S deficiency and PH. These findings carry broad translational implications for defining the metabolic origins of PH and potentially other metabolic diseases sharing similar underpinnings.
Collapse
Affiliation(s)
- Kevin White
- Divisions of Cardiovascular Medicine and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yu Lu
- Divisions of Cardiovascular Medicine and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sofia Annis
- Divisions of Cardiovascular Medicine and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew E Hale
- Divisions of Cardiovascular Medicine and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - James E Dahlman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Craig Hemann
- The Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Alexander R Opotowsky
- Divisions of Cardiovascular Medicine and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara O Vargas
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ivan Rosas
- Division of Pulmonary/Critical Care Medicine, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Mark A Perrella
- Division of Pulmonary/Critical Care Medicine, Department of Medicine, Harvard Medical School, Boston, MA, USA Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Juan C Osorio
- Division of Pulmonary/Critical Care Medicine, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kathleen J Haley
- Division of Pulmonary/Critical Care Medicine, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Brian B Graham
- Program in Translational Lung Research, University of Colorado, Denver, Aurora, CO, USA
| | - Rahul Kumar
- Program in Translational Lung Research, University of Colorado, Denver, Aurora, CO, USA
| | - Rajan Saggar
- Departments of Medicine and Pathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rajeev Saggar
- Department of Cardiothoracic Surgery, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - W Dean Wallace
- Departments of Medicine and Pathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - David J Ross
- Departments of Medicine and Pathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Omar F Khan
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew Bader
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bernadette R Gochuico
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | - Haydn M Prosser
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Daniel G Anderson
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jay L Zweier
- The Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Laurence A Bindoff
- Department of Clinical Medicine, University of Bergen, Bergen, Norway Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - David Systrom
- Division of Pulmonary/Critical Care Medicine, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Aaron B Waxman
- Division of Pulmonary/Critical Care Medicine, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Richard C Jin
- Divisions of Cardiovascular Medicine and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen Y Chan
- Divisions of Cardiovascular Medicine and Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
140
|
Shi L, Liao J, Liu B, Zeng F, Zhang L. Mechanisms and therapeutic potential of microRNAs in hypertension. Drug Discov Today 2015; 20:1188-204. [PMID: 26004493 DOI: 10.1016/j.drudis.2015.05.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/27/2015] [Accepted: 05/14/2015] [Indexed: 01/08/2023]
Abstract
Hypertension is the major risk factor for the development of stroke, coronary artery disease, heart failure and renal disease. The underlying cellular and molecular mechanisms of hypertension are complex and remain largely elusive. MicroRNAs (miRNAs) are short, noncoding RNA fragments of 22-26 nucleotides and regulate protein expression post-transcriptionally by targeting the 3'-untranslated region of mRNA. A growing body of recent research indicates that miRNAs are important in the pathogenesis of arterial hypertension. Herein, we summarize the current knowledge regarding the mechanisms of miRNAs in cardiovascular remodeling, focusing specifically on hypertension. We also review recent progress of the miRNA-based therapeutics including pharmacological and nonpharmacological therapies (such as exercise training) and their potential applications in the management of hypertension.
Collapse
Affiliation(s)
- Lijun Shi
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China.
| | - Jingwen Liao
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China
| | - Bailin Liu
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China
| | - Fanxing Zeng
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China
| | - Lubo Zhang
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
141
|
McLendon JM, Joshi SR, Sparks J, Matar M, Fewell JG, Abe K, Oka M, McMurtry IF, Gerthoffer WT. Lipid nanoparticle delivery of a microRNA-145 inhibitor improves experimental pulmonary hypertension. J Control Release 2015; 210:67-75. [PMID: 25979327 DOI: 10.1016/j.jconrel.2015.05.261] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/04/2015] [Accepted: 05/09/2015] [Indexed: 12/14/2022]
Abstract
Therapies that exploit RNA interference (RNAi) hold great potential for improving disease outcomes. However, there are several challenges that limit the application of RNAi therapeutics. One of the most important challenges is effective delivery of oligonucleotides to target cells and reduced delivery to non-target cells. We have previously developed a functionalized cationic lipopolyamine (Star:Star-mPEG-550) for in vivo delivery of siRNA to pulmonary vascular cells. This optimized lipid formulation enhances the retention of siRNA in mouse lungs and achieves significant knockdown of target gene expression for at least 10days following a single intravenous injection. Although this suggests great potential for developing lung-directed RNAi-based therapies, the application of Star:Star-mPEG mediated delivery of RNAi based therapies for pulmonary vascular diseases such as pulmonary arterial hypertension (PAH) remains unknown. We identified differential expression of several microRNAs known to regulate cell proliferation, cell survival and cell fate that are associated with development of PAH, including increased expression of microRNA-145 (miR-145). Here we test the hypothesis that Star:Star-mPEG mediated delivery of an antisense oligonucleotide against miR-145 (antimiR-145) will improve established PAH in rats. We performed a series of experiments testing the in vivo distribution, toxicity, and efficacy of Star:Star-mPEG mediated delivery of antimiR-145 in rats with Sugen-5416/hypoxia induced PAH. We showed that after subchronic therapy of three intravenous injections over 5weeks at 2mg/kg, antimiR-145 accumulated in rat lung tissue and reduced expression of endogenous miR-145. Using a novel in situ hybridization approach, we demonstrated substantial distribution of antimiR-145 in the lungs as well as the liver, kidney, and spleen. We assessed toxic effects of Star:Star-mPEG/antimiR-145 with serial complete blood counts of leukocytes and serum metabolic panels, gross pathology, and histopathology and did not detect significant off-target effects. AntimiR-145 reduced the degree of pulmonary arteriopathy, reduced the severity of pulmonary hypertension, and reduced the degree of cardiac dysfunction. The results establish effective and low toxicity of lung delivery of a miRNA-145 inhibitor using functionalized cationic lipopolyamine nanoparticles to repair pulmonary arteriopathy and improve cardiac function in rats with severe PAH.
Collapse
Affiliation(s)
- Jared M McLendon
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA; Center for Lung Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA.
| | - Sachindra R Joshi
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA; Center for Lung Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA
| | - Jeff Sparks
- Celsion-EGEN, 601 Genome Way, Huntsville, AL 35806, USA
| | - Majed Matar
- Celsion-EGEN, 601 Genome Way, Huntsville, AL 35806, USA
| | | | - Kohtaro Abe
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA
| | - Masahiko Oka
- Department of Internal Medicine, University of South Alabama College of Medicine, Mobile, AL 36688, USA; Center for Lung Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA
| | - Ivan F McMurtry
- Department of Pharmacology, University of South Alabama College of Medicine, Mobile, AL 36688, USA; Department of Internal Medicine, University of South Alabama College of Medicine, Mobile, AL 36688, USA; Center for Lung Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA
| | - William T Gerthoffer
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA; Center for Lung Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA
| |
Collapse
|
142
|
Brazil DP, Church RH, Surae S, Godson C, Martin F. BMP signalling: agony and antagony in the family. Trends Cell Biol 2015; 25:249-64. [DOI: 10.1016/j.tcb.2014.12.004] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 12/01/2014] [Accepted: 12/02/2014] [Indexed: 01/14/2023]
|
143
|
Fox CS, Hall JL, Arnett DK, Ashley EA, Delles C, Engler MB, Freeman MW, Johnson JA, Lanfear DE, Liggett SB, Lusis AJ, Loscalzo J, MacRae CA, Musunuru K, Newby LK, O'Donnell CJ, Rich SS, Terzic A. Future translational applications from the contemporary genomics era: a scientific statement from the American Heart Association. Circulation 2015; 131:1715-36. [PMID: 25882488 DOI: 10.1161/cir.0000000000000211] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The field of genetics and genomics has advanced considerably with the achievement of recent milestones encompassing the identification of many loci for cardiovascular disease and variable drug responses. Despite this achievement, a gap exists in the understanding and advancement to meaningful translation that directly affects disease prevention and clinical care. The purpose of this scientific statement is to address the gap between genetic discoveries and their practical application to cardiovascular clinical care. In brief, this scientific statement assesses the current timeline for effective translation of basic discoveries to clinical advances, highlighting past successes. Current discoveries in the area of genetics and genomics are covered next, followed by future expectations, tools, and competencies for achieving the goal of improving clinical care.
Collapse
|
144
|
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease without effective treatment. Despite decades of research and the development of novel treatments, PAH remains a fatal disease, suggesting an urgent need for better understanding of the pathogenesis of PAH. Recent studies suggest that microRNAs (miRNAs) are dysregulated in patients with PAH and in experimental pulmonary hypertension. Furthermore, normalization of a few miRNAs is reported to inhibit experimental pulmonary hypertension. We have reviewed the current knowledge about miRNA biogenesis, miRNA expression pattern, and their roles in regulation of pulmonary artery smooth muscle cells, endothelial cells, and fibroblasts. We have also identified emerging trends in our understanding of the role of miRNAs in the pathogenesis of PAH and propose future studies that might lead to novel therapeutic strategies for the treatment of PAH.
Collapse
Affiliation(s)
- Guofei Zhou
- 1 Department of Pediatrics, University of Illinois at Chicago; and
| | | | | |
Collapse
|
145
|
Rothman AMK, Chico TJA, Lawrie A. MicroRNA in pulmonary vascular disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 124:43-63. [PMID: 24751426 DOI: 10.1016/b978-0-12-386930-2.00003-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
MicroRNA (miRNA) are short noncoding RNA that regulate gene expression by inhibiting translation or promoting degradation of target mRNA. miRNA are key regulators of a wide range of cellular processes and their discovery has revolutionized our understanding of gene regulatory networks. Pulmonary arterial hypertension (PAH) is a debilitating and fatal disease characterized by remodeling of pulmonary arteries and right heart failure. Factors including sustained pulmonary vasoconstriction, inflammation, and altered cellular signaling pathways drive disease through pulmonary artery endothelial dysfunction, smooth muscle cell proliferation, and the recruitment of circulating cells. miRNA have been shown to regulate many of the key drivers of pathology, yet the role of only a limited number of miRNA has been recognized in PAH. Investigation of the diverse regulatory functions of miRNA offers the potential to further understanding of the cellular pathology of PAH and to provide much needed diagnostic and therapeutic strategies. This review focuses on recent advances in the investigation of miRNA in PAH.
Collapse
Affiliation(s)
- Alex M K Rothman
- Department of Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom
| | - Timothy J A Chico
- Department of Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom
| | - Allan Lawrie
- Department of Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
146
|
A novel minimally-invasive method to sample human endothelial cells for molecular profiling. PLoS One 2015; 10:e0118081. [PMID: 25679506 PMCID: PMC4332500 DOI: 10.1371/journal.pone.0118081] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 01/05/2015] [Indexed: 01/05/2023] Open
Abstract
Objective The endothelium is a key mediator of vascular homeostasis and cardiovascular health. Molecular research on the human endothelium may provide insight into the mechanisms underlying cardiovascular disease. Prior methodology used to isolate human endothelial cells has suffered from poor yields and contamination with other cell types. We thus sought to develop a minimally invasive technique to obtain endothelial cells derived from human subjects with higher yields and purity. Methods Nine healthy volunteers underwent endothelial cell harvesting from antecubital veins using guidewires. Fluorescence-activated cell sorting (FACS) was subsequently used to purify endothelial cells from contaminating cells using endothelial surface markers (CD34 / CD105 / CD146) with the concomitant absence of leukocyte and platelet specific markers (CD11b / CD45). Endothelial lineage in the purified cell population was confirmed by expression of endothelial specific genes and microRNA using quantitative polymerase chain reaction (PCR). Results A median of 4,212 (IQR: 2161 – 6583) endothelial cells were isolated from each subject. Quantitative PCR demonstrated higher expression of von Willebrand Factor (vWF, P<0.001), nitric oxide synthase 3 (NOS3, P<0.001) and vascular cell adhesion molecule 1 (VCAM-1, P<0.003) in the endothelial population compared to similarly isolated leukocytes. Similarly, the level of endothelial specific microRNA-126 was higher in the purified endothelial cells (P<0.001). Conclusion This state-of-the-art technique isolates human endothelial cells for molecular analysis in higher purity and greater numbers than previously possible. This approach will expedite research on the molecular mechanisms of human cardiovascular disease, elucidating its pathophysiology and potential therapeutic targets.
Collapse
|
147
|
Lee A, McLean D, Choi J, Kang H, Chang W, Kim J. Therapeutic implications of microRNAs in pulmonary arterial hypertension. BMB Rep 2015; 47:311-7. [PMID: 24755557 PMCID: PMC4163875 DOI: 10.5483/bmbrep.2014.47.6.085] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Indexed: 12/29/2022] Open
Abstract
microRNAs (miRNAs) are a class of small, non-coding RNAs that play critical posttranscriptional regulatory roles typically through targeting of the 3'-untranslated region of messenger RNA (mRNA). Mature miRNAs are known to be involved in global cellular processes, such as differentiation, proliferation, apoptosis, and organogenesis, due to their capacity to target multiple mRNAs. Thus, imbalances in the expression and/or activity of miRNAs are involved in the pathogenesis of numerous diseases, including pulmonary arterial hypertension (PAH). PAH is a progressive disease characterized by vascular remodeling due to excessive proliferation of pulmonary artery endothelial cells (PAECs) and pulmonary artery smooth muscle cells (PASMCs). Recently, studies have evaluated the roles of miRNAs involved in the pathogenesis of PAH in these pulmonary vascular cells. This review provides an overview of recent discoveries on the role of miRNAs in the pathogenesis of PAH and discusses the potential for miRNAs as therapeutic targets and biomarkers of PAH. [BMB Reports 2014; 47(6): 311-317]
Collapse
Affiliation(s)
- Aram Lee
- Department of Life Systems, Sookmyung Women's University, Seoul 140-742, Korea
| | - Danielle McLean
- Cardiovascular Research Institute, University of Vermont, 208 South Park Drive, Colchester, VT 05446, USA
| | - Jihea Choi
- Department of Life Systems, Sookmyung Women's University, Seoul 140-742, Korea
| | - Hyesoo Kang
- Department of Life Systems, Sookmyung Women's University, Seoul 140-742, Korea
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Busan 609-735, Korea
| | - Jongmin Kim
- Department of Life Systems, Sookmyung Women's University, Seoul 140-742, Korea
| |
Collapse
|
148
|
Bertero T, Cottrill K, Krauszman A, Lu Y, Annis S, Hale A, Bhat B, Waxman AB, Chau BN, Kuebler WM, Chan SY. The microRNA-130/301 family controls vasoconstriction in pulmonary hypertension. J Biol Chem 2015; 290:2069-85. [PMID: 25505270 PMCID: PMC4303661 DOI: 10.1074/jbc.m114.617845] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/26/2014] [Indexed: 12/19/2022] Open
Abstract
Pulmonary hypertension (PH) is a complex disorder, spanning several known vascular cell types. Recently, we identified the microRNA-130/301 (miR-130/301) family as a regulator of multiple pro-proliferative pathways in PH, but the true breadth of influence of the miR-130/301 family across cell types in PH may be even more extensive. Here, we employed targeted network theory to identify additional pathogenic pathways regulated by miR-130/301, including those involving vasomotor tone. Guided by these predictions, we demonstrated, via gain- and loss-of-function experimentation in vitro and in vivo, that miR-130/301-specific control of the peroxisome proliferator-activated receptor γ regulates a panel of vasoactive factors communicating between diseased pulmonary vascular endothelial and smooth muscle cells. Of these, the vasoconstrictive factor endothelin-1 serves as an integral point of communication between the miR-130/301-peroxisome proliferator-activated receptor γ axis in endothelial cells and contractile function in smooth muscle cells. Thus, resulting from an in silico analysis of the architecture of the PH disease gene network coupled with molecular experimentation in vivo, these findings clarify the expanded role of the miR-130/301 family in the global regulation of PH. They further emphasize the importance of molecular cross-talk among the diverse cellular populations involved in PH.
Collapse
Affiliation(s)
- Thomas Bertero
- From the Divisions of Cardiovascular and Network Medicine and
| | | | - Adrienn Krauszman
- the Keenan Research Centre for Biomedical Science of St. Michael's, University of Toronto, Toronto, Ontario M5R 0A3, Canada
| | - Yu Lu
- From the Divisions of Cardiovascular and Network Medicine and
| | - Sofia Annis
- From the Divisions of Cardiovascular and Network Medicine and
| | - Andrew Hale
- From the Divisions of Cardiovascular and Network Medicine and
| | | | - Aaron B Waxman
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - B Nelson Chau
- Regulus Therapeutics, San Diego, California 92121, and
| | - Wolfgang M Kuebler
- the Keenan Research Centre for Biomedical Science of St. Michael's, University of Toronto, Toronto, Ontario M5R 0A3, Canada, the Department of Physiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Stephen Y Chan
- From the Divisions of Cardiovascular and Network Medicine and
| |
Collapse
|
149
|
Lundstrom K. Personalized Medicine and Epigenetic Drug Development. PERSONALIZED EPIGENETICS 2015:369-386. [DOI: 10.1016/b978-0-12-420135-4.00013-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
150
|
Boucherat O, Potus F, Bonnet S. microRNA and Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 888:237-52. [PMID: 26663186 DOI: 10.1007/978-3-319-22671-2_12] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a lethal vasculopathy associated with complex etiology that involves remodeling of distal pulmonary arteries leading to elevation of pulmonary vascular resistance. This process results in right ventricular (RV) hypertrophy and ultimately RV failure. In addition, PAH is associated with systemic impairment in the skeletal muscle contributing to exercise intolerance. It has only been a few decades since microRNAs (miRNAs) have been implied in the development and progression of PAH regarding every organ affected by the disease. Indeed, impairment of miRNA's expression has been involved in vascular cell remodeling processes such as adventitial fibroblast (AdvFB) migration; pulmonary arterial smooth muscle cell (PASMC) proliferation and pulmonary arterial endothelial cell (PAEC) dysfunction observed in PAH. At the molecular level miRNAs have been described in the control of ion channels and mitochondrial function as well as the regulation of the BMPR2 signaling pathways contributing to PAH lung impairment. Recently miRNAs have also been specifically implicated in RV dysfunction and systemic angiogenic impairment, observed in PAH. In this chapter, we will summarize the knowledge on miRNA in PAH and highlight their crucial role in the etiology of this disease.
Collapse
Affiliation(s)
- Olivier Boucherat
- Pulmonary Hypertension Research Group of the University Institute of Cardiology and Pneumology, Québec Research Center, Laval University, Quebec City, QC, Canada
| | - François Potus
- Pulmonary Hypertension Research Group of the Quebec Heart and Lung Institute, Québec Research Center, Laval University, Quebec City, QC, Canada
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group of the Quebec Heart and Lung Institute, Québec Research Center, Laval University, Quebec City, QC, Canada.
| |
Collapse
|